12
Charles Darwin University Circumsporozoite-specific T cell responses in children vaccinated with RTS,S/AS01 E and protection against P falciparum clinical malaria Olotu, Ally; Moris, Philippe; Mwacharo, Jedidah; Vekemans, Johan; Kimani, Domtila; Janssens, Michel; Kai, Oscar; Jongert, Erik; Lievens, Marc; Leach, Amanda; Villafana, Tonya; Savarese, Barbara; Marsh, Kevin; Cohen, Joe; Bejon, Philip Published in: PLoS One DOI: 10.1371/journal.pone.0025786 Published: 01/01/2011 Document Version Publisher's PDF, also known as Version of record Link to publication Citation for published version (APA): Olotu, A., Moris, P., Mwacharo, J., Vekemans, J., Kimani, D., Janssens, M., Kai, O., Jongert, E., Lievens, M., Leach, A., Villafana, T., Savarese, B., Marsh, K., Cohen, J., & Bejon, P. (2011). Circumsporozoite-specific T cell responses in children vaccinated with RTS,S/AS01 E and protection against P falciparum clinical malaria. PLoS One, 6(10), 1-11. [e25786]. https://doi.org/10.1371/journal.pone.0025786 General rights Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. • Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain • You may freely distribute the URL identifying the publication in the public portal Take down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Download date: 29. Apr. 2020

Charles Darwin University Circumsporozoite-specific T cell ... · Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01 E and Protection against P falciparumClinical

  • Upload
    others

  • View
    8

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Charles Darwin University Circumsporozoite-specific T cell ... · Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01 E and Protection against P falciparumClinical

Charles Darwin University

Circumsporozoite-specific T cell responses in children vaccinated with RTS,S/AS01 Eand protection against P falciparum clinical malaria

Olotu, Ally; Moris, Philippe; Mwacharo, Jedidah; Vekemans, Johan; Kimani, Domtila;Janssens, Michel; Kai, Oscar; Jongert, Erik; Lievens, Marc; Leach, Amanda; Villafana, Tonya;Savarese, Barbara; Marsh, Kevin; Cohen, Joe; Bejon, PhilipPublished in:PLoS One

DOI:10.1371/journal.pone.0025786

Published: 01/01/2011

Document VersionPublisher's PDF, also known as Version of record

Link to publication

Citation for published version (APA):Olotu, A., Moris, P., Mwacharo, J., Vekemans, J., Kimani, D., Janssens, M., Kai, O., Jongert, E., Lievens, M.,Leach, A., Villafana, T., Savarese, B., Marsh, K., Cohen, J., & Bejon, P. (2011). Circumsporozoite-specific T cellresponses in children vaccinated with RTS,S/AS01 E and protection against P falciparum clinical malaria. PLoSOne, 6(10), 1-11. [e25786]. https://doi.org/10.1371/journal.pone.0025786

General rightsCopyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright ownersand it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights.

• Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain • You may freely distribute the URL identifying the publication in the public portal

Take down policyIf you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediatelyand investigate your claim.

Download date: 29. Apr. 2020

Page 2: Charles Darwin University Circumsporozoite-specific T cell ... · Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01 E and Protection against P falciparumClinical

Circumsporozoite-Specific T Cell Responses in ChildrenVaccinated with RTS,S/AS01E and Protection againstP falciparum Clinical MalariaAlly Olotu1, Philippe Moris2, Jedidah Mwacharo1, Johan Vekemans2, Domtila Kimani1, Michel Janssens2,

Oscar Kai1, Erik Jongert2, Marc Lievens2, Amanda Leach2, Tonya Villafana3,4, Barbara Savarese3, Kevin

Marsh1,5, Joe Cohen2, Philip Bejon1,5*

1 Kenya Medical Research Institute/ Wellcome Trust Programme, Centre for Geographic Medicine Research, Coast, Kilifi, Kenya, 2 GlaxoSmithKline Biologicals, Rixensart,

Belgium, 3 PATH Malaria Vaccine Initiative (MVI), Bethesda, Maryland, United States of America, 4 MedImmune, LLC, Gaithersburg, Maryland, United States of America,

5 Centre for Clinical Vaccinology and Tropical Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom

Abstract

Background: RTS,S/AS01E is the lead candidate pre-erythrocytic malaria vaccine. In Phase IIb field trials the safety profile wasacceptable and the efficacy was 53% (95%CI 31%–72%) for protecting children against clinical malaria caused by P.falciparum. We studied CS-specific T cell responses in order to identify correlates of protection.

Methods and Findings: We used intracellular cytokine staining (for IL2, IFNc, and TNFa), ex-vivo ELISPOTs (IFNc and IL2) andIFNc cultured ELISPOT assays to characterize the CS-specific cellular responses in 407 children (5–17 months of age) in aphase IIb randomized controlled trial of RTS,S/AS01E (NCT00380393). RTS,S/ AS01E vaccinees had higher frequencies of CS-specific CD4+ T cells producing IFNc, TNFa or IL2 compared to control vaccinees. In a multivariable analysis TNFa+ CD4+ Tcells were independently associated with a reduced risk for clinical malaria among RTS,S/AS01E vaccinees (HR = 0.64, 95%CI0.49–0.86, p = 0.002). There was a non-significant tendency towards reduced risk among control vaccinees (HR = 0.80, 95%CI0.62–1.03, p = 0.084), albeit with lower CS-specific T cell frequencies and higher rates of clinical malaria. When data fromboth RTS,S/AS01E vaccinees and control vaccinees were combined (with adjusting for vaccination group), the HR was 0.74(95%CI 0.62–0.89, p = 0.001). After a Bonferroni correction for multiple comparisons (n-18), the finding was still significant atp = 0.018. There was no significant correlation between cultured or ex vivo ELISPOT data and protection from clinical malaria.The combination of TNFa+ CD4+ T cells and anti-CS antibody statistically accounted for the protective effect of vaccinationin a Cox regression model.

Conclusions: RTS,S/AS01E induces CS-specific Th1 T cell responses in young children living in a malaria endemic area. Thecombination of anti-CS antibody concentrations titers and CS-specific TNFa+ CD4+ T cells could account for the level ofprotection conferred by RTS,S/AS01E. The correlation between CS-specific TNFa+ CD4+ T cells and protection needsconfirmation in other datasets.

Citation: Olotu A, Moris P, Mwacharo J, Vekemans J, Kimani D, et al. (2011) Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01E

and Protection against P falciparum Clinical Malaria. PLoS ONE 6(10): e25786. doi:10.1371/journal.pone.0025786

Editor: Lorenz von Seidlein, Menzies School of Health Research, Australia

Received July 11, 2011; Accepted September 9, 2011; Published October 6, 2011

Copyright: � 2011 Olotu et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permitsunrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Funding: This project has been funded by PATH Malaria Vaccine Initiative (MVI). Prof. Marsh is supported by the Wellcome Trust. Philip Bejon is supported by theBiomedical Research Centre in Oxford. Two authors (TV and BS) are MVI employees and were involved in trial design and writing the report. Dr. Villafana was anemployee of MVI throughout the duration of the study, but has since moved to MedImmune, LLC. MedImmune, LLC had no role in the study design, datacollection, analysis, decision to publish, preparation of the manuscript or any other aspect of the study. The Wellcome Trust and Biomedical Research Centre hadno role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. MVI had no other role in data collection or analysis, andthe decision to publish was covered by a pre-study agreement.

Competing Interests: Dr. Leach, Mr. Lievens, Dr. Vekemans, Dr. Jongert, and Dr. Cohen report being employees of GlaxoSmithKline Biologicals; Dr. Leach,Dr. Vekemans and Dr. Cohen report owning shares in GlaxoSmithKline. Dr. Cohen reports being listed as an inventor of patents covering the RTS,S malaria vaccinecandidate, which include patents and patent applications related to International Patent Application Publication Nos. WO93/10152, WO2006/029887, WO2007/003384, WO2009/021931 and WO2009/080715, the rights to which have been assigned to GlaxoSmithKline Biologicals. Ms. Savarese, and Dr. Villafana reportbeing employees of MVI, which supports the development and testing of several malaria vaccines (i.e. Ad26/35-CSP (Crucell), PvRII (ICGEB), whole irradiatedsporozoites (Sanaria), MSP2 (LaTrobe/QIMR), AMA1 (NIH/QIMR) and RTS,S/AS01). Dr. Villafana was an employee of MVI throughout the duration of the study, buthas since moved to MedImmune, LLC. No other potential conflict of interest relevant to this article was reported. This does not alter the authors’ adherence to allthe PLoS ONE policies on sharing data and materials.

* E-mail: [email protected]

Introduction

RTS,S is the lead candidate pre-erythrocytic malaria vaccine

[1]. The vaccine antigen consists of 19 copies of the central

tandem repeats and C-terminal region of the P. falciparum

circumsporozoite protein (CS) fused to hepatitis B surface antigen

(HBsAg), and co-expressed with unfused HBsAg in Saccharomyces

cerevisiae cells. The two proteins spontaneously assemble in the

yeast cells to form virus-like particles. The RTS,S antigen has been

tested with two different alternative Adjuvant Systems: AS02 or

PLoS ONE | www.plosone.org 1 October 2011 | Volume 6 | Issue 10 | e25786

Page 3: Charles Darwin University Circumsporozoite-specific T cell ... · Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01 E and Protection against P falciparumClinical

AS01. Both Adjuvant Systems contain the immunostimulants

monophosphoryl lipid A (MPLH) and QS21, formulated either

with an oil-in-water emulsion (AS02) or with liposomes (AS01).

Formulated in either Adjuvant System, the RTS,S antigen

induces high concentrations of anti-circumsporozoite protein (CS)

antibodies [2,3,4,5,6,7]. Correlations between anti-CS concentra-

tions and protection against infection were statistically significant

on experimental challenge with P. falciparum in malaria naı̈ve

adults [7], of borderline significance on natural challenge of semi-

immune adults [4], and significant on natural challenge of children

in a malaria endemic area [8]. Anti-CS titers did not correlate with

protection against clinical malaria episodes in children [4,9], but

we recently identified a non-linear relationship between concur-

rent (rather than peak) anti-CS titers and protection from clinical

malaria in children [10].

CD4+ T cell responses to pre-erythrocytic antigens prevent

intra-hepatocytic parasites developing in both human and mouse

studies [11,12]. Potential mechanisms include TNFa induced

apoptosis [13] or inhibition of parasite growth [14] and IFNcinduced NO production [15]. RTS,S-induced cell mediated

immune responses have been assessed using proliferation assays,

cytokine production on cell culture, intracellular cytokine staining

and flow-cytometry, and ex-vivo and cultured ELISPOT assays

[16,17].

RTS,S/AS immunization induces a CD4+ T cell response but

little or no detectable CD8+ T cell response [7,18,19,20,21]. Sun

et al observed IFNc-producing CD8+ T cells, but only after cells

were stimulated for 10–14 days in vitro [22]. Barbosa et al reported

CD8+ T cell responses after 42 hours in vitro stimulation on

comparing RTS,S/AS02 vaccinees with control vaccinees at 10

weeks, but not at 4 weeks, post immunization [23].

The frequency of poly-functional CD4+ T cells identified by

intracellular cytokine staining (ICS) correlated with protection

from P. falciparum infection after experimental challenge in adults

[7,24]. In a field study, Reece et al reported a correlation between

protection against re-infection and cultured IFNc ELISPOT

assays using a single conserved T cell epitope from the CS protein

[20]. However, this analysis was not adjusted for anti-CS titers,

and did not include ICS studies. A borderline correlation between

single cytokine ICS results and protection from P. falciparum

infection was shown in a field study in infants [23].

In order to examine associations with protection against clinical

malaria, we assessed the CS-specific cellular immune responses in

447 children using ICS, ex vivo IFNc and IL2 ELISPOT, and

cultured IFNc ELISPOT assays in a phase II b randomized

clinical trial of RTS,S/AS01E versus control, in which we

observed 53% (95%CI 31%–72%) protection against clinical

malaria [25]. The blood volumes sampled in children prevented us

from using an ICS assay previously reported in adult studies [7],

but a whole blood ICS assay requiring smaller blood volumes has

been developed and used in two phase II trials in Ghana [26] and

Gabon [27]. These studies showed that the vaccine induced CD4+IL2, TNFa or IFNc producing cells, but CD40L was not

detectable using the whole blood assay for children in Sub-

Saharan Africa. We therefore did not include CD40L staining in

the assay for our study.

The qualification of correlates of immunity and surrogates of

protection has been recently reviewed [28,29]. The Prentice

criteria require that: a) vaccination predicts protection; b)

vaccination predicts the potential surrogate; c) the surrogate

predicts protection among vaccinees and d) that the surrogate

accounts for all the effect of vaccination [30]. If vaccination is an

independent predictor of outcome after including the potential

surrogate in the analysis, this suggests that other mechanisms are

involved. On the other hand, if including the potential surrogate in

analysis removes vaccination as a predictor, this is consistent with

the effect of vaccination being mediated by the surrogate marker.

Methods

The study protocol and its amendments received ethical and

scientific approval from Kenya Medical Research Institute

National Ethics Committee, National Institute for Medical

Research of Tanzania, the Oxford Tropical Research Ethics

Committee, the London School of Hygiene and Tropical

Medicine Ethics committee and the Western Institutional Review

Board in Seattle. The study was conducted in accordance with the

Helsinki Declaration of 1964 (revised 1996) and Good Clinical

Practice guidelines and was overseen by an Independent data

monitoring committee and local safety monitors. Written informed

consent was obtained using approved Swahili or Giriama consent

forms. Illiterate parents thumb printed the consent form which

was countersigned by an independent, literate witness.

We conducted a randomized controlled trial to evaluate the

efficacy and safety of RTS,S/AS01E against clinical malaria

episodes due to P. falciparum infection in Kilifi, Kenya and

Korogwe, Tanzania. There were 894 children between the two

sites, of which the 447 children enrolled in Kilifi were assessed for

vaccine induced cellular immunity using ICS and ELISPOT.

Details on randomization, immunization and surveillance have

been published previously [25]. In Kilifi, 447 children 5–17 months

old were randomized and received either RTS,S/AS01E or rabies

vaccine in a 1:1 ratio according to 0, 1, 2 month schedule. Both

vaccines were given intramuscularly in the left deltoid. The primary

end point was clinical malaria, defined as the presence of fever

(axillary temperature $37.5uC) and P. falciparum parasitaemia

$2500/mL. Active and passive surveillance for malaria was

conducted by field workers and study personnel at local dispensaries.

Children were vaccinated between March and August 2007.

Blood was taken for immunological studies before vaccination, one

month post dose 3, then on March 2008 irrespective of the time of

recruitment (i.e. between 4 and 10 months post dose 3, mean 8

months), 12 months post dose 3 and in October 2008 irrespective

of time of recruitment, (i.e. between 12 and 18 months post dose 3,

mean 15 months). Peak malaria transmission was between May

and August 2008.

CS antibody measurementAntibodies to the P. falciparum circumsporozoite protein (CS) tandem

repeat epitope were assessed by ELISA at the Center for Vaccinology,

Ghent University Hosptial, Belgium. Results were reported in EU/

mL. Plates were adsorbed with the recombinant antigen R32LR that

contained the sequence [NVDP(NANP)15]2LR [31].

PeptidesA set of 32 15-mer, peptides were used, overlapping by 11

amino acids to cover the full length of the CS antigen used in the

vaccine (3D7 strain). All these peptides were used in a single pool

for the ICS studies, but they were divided into three pools for

ELISPOT studies, namely; a) the conserved region including the

NANP repeats, b) the variant TH2R region and c) the variant

TH3R region and conserved CS.T3T region (Table 1).

ELISPOT assaysPeripheral blood mononuclear cells (PBMC) were separated

and incubated in RPMI medium (Sigma-Aldrich) with 10%

Human AB serum. We used Millipore MAIP S45 plates and

MabTech antibodies for ELISPOT assays according to the

T Cell Immunity and RTS,S/AS01E

PLoS ONE | www.plosone.org 2 October 2011 | Volume 6 | Issue 10 | e25786

Page 4: Charles Darwin University Circumsporozoite-specific T cell ... · Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01 E and Protection against P falciparumClinical

manufacturer’s instructions. For ex vivo ELISPOT assays (IFNcand IL2), 26105 per well of freshly isolated PBMCs were

incubated in 100ml final volume at 2.5 mg/ml circumsporozoite

antigen peptides (see table 1) for 18–20 hours before developing

the plates. The positive and negative controls were 20 mg/ml

Phytohaemaglutinin (Sigma-Aldrich) and media alone, respective-

ly. For cultured ELISPOTs, 1x106 PBMC were incubated in

0.5 mls of 10mg/ml/peptide of pooled peptides in a 24-well plate.

On days 3 and 7, 250ml of culture supernatant was replaced with

250ml culture medium containing 20 IU/ml recombinant IL 2.

On day 9, the cells were washed three times and left overnight

before an ELISPOT assay (IFNc only) was done according to the

method used for ex vivo ELISPOTs. Spot forming cell numbers

were counted by ELISPOT plate reader (Autoimmun Diagnos-

tika, version 3.0).

ELISPOT analysisELISPOT wells were assayed in duplicate, and the final result

was the mean of two wells. The negative control well result was

subtracted from each peptide well. ELISPOTs failed quality

control if the negative control well had more than 25 spots or the

positive control had less than 50 spots. The results from the three

peptide pools were added to calculate total responses. Results are

presented as number of spots per million incubated PBMC.

Whole blood ICS assayWhole blood was stimulated in Kilifi within 2 hours of being

drawn. 350 ml of whole blood plus 100 ml of phosphate buffered

saline (PBS) was incubated in three different 15 ml Falcon tube,

with 1 mg/ml of anti-CD28 anti-CD49d monoclonal antibodies

(supplied by BD). After 2 hours, Brefeldin A was added to a final

concentration of 1 mg/ml and incubation was continued overnight

at 37 ˚C 6 1- CO2 5 to 7%. EDTA was then added to a final

concentration at 5 mM, and after 15 minutes 1 ml FACS lysing

solution (BD). The positive control was stimulated using

staphylococcal enterotoxin B (SEB) and negative control was

PBS without peptides. Circumsporozoite antigen peptides were

added to the third tube to a final concentration of 1 mg/ml (see

Table 1). The cells were then washed in PBS and re-suspended in

PBS with 10% DMSO and stored at –70 ˚C for transport to GSK

in Rixensart. In GSK, cells were thawed, washed and stained with

alexa-fluor 700 conjugated anti-CD3 (Pharmingen), peridinin-

chlorophyll (PerCP)-conjugated anti-CD4 (BD Biosciences) and

allophycocyanin (APC)-H7 conjugated anti-CD8 antibodies (BD

Biosciences). Cells were fixed and permeabilized using the

Cytofix/Cytoperm buffer kit (Pharmingen), and stained with

APC conjugated anti-IL-2 (Pharmingen), fluorescein-isothiocya-

nate (FITC)-conjugated anti-IFN-c (Pharmingen) and phycoery-

thrin (PE) cyanin–7 (Cy7)-conjugated anti-TNFa (Pharmingen).

Cells were washed, re-suspended in fetal-calf-serum (FCS)-

containing phosphate buffered saline (PBS) and analyzed on a

BDTM LSR II flow cytometer (BD Biosciences). Events were

counted using the automatic gating on the FACSDiva software

(BD Biosciences). Conventional rules were used to gate on single

cells, then the lymphocyte subset based on forward and side

scatter. CD3 and CD4/CD8 positive cells and then cytokine

expression was classified into positive/negative using FACSDiva

software. An example of the output with gating shown is given in

Supporting Information S1. We required at least 10,000 CD4 +events and 5,000 CD8+ events. Acquisition was stopped when

75,000 CD4+ events had been acquired, and we acquired more

than 50,000 CD4+ events for the majority of samples (.90%).

Results from antigen-stimulated cultures were not excluded from

analysis on the basis of positive/negative control results, in the

absence of established criteria. Data are represented as background

subtracted CS-specific events per million CD4+ or CD8+ T cells.

Assays were conducted according to sample availability, since

blood samples were limited to 5 mls. In order of priority, the

assays conducted were; ICS, IFNc ex vivo ELISPOT, IL2 ex vivo

ELISPOT and cultured ELISPOT. Samples were processed

within 3 hours of being taken. ICS samples were stored for 3 to

4 months at –70 ˚C before staining. The samples were processed

during the double-blind phase of the study.

Statistical analysisGeometric mean responses are calculated and a Student’s T test

was performed on log-transformed values to compare between

vaccination groups. A paired T test on log-transformed values was

used to compare time-courses, and correlations between assays

were examined using Pearson’s product moment calculation on

Table 1. Peptide pools.

Peptide Sequence

NANP and conserved region peptides pool

Pept 1 MMAP DPNANPNANPN

Pept 2 NANP NANPNANPNAN

Pept 3 DPNA NPNANPNKNNQ

Pept 4 NPNA NPNKNNQGNGQ

Pept 5 NPNK NNQGNGQGHNM

Pept 6 NNQG NGQGHNMPNDP

Pept 7 NGQG HNMPNDPNRNV

Pept 8 HNMP NDPNRNVDENA

Pept 9 NDPN RNVDENANANS

Pept 10 RNVD ENANANSAVKN

Pept 11 ENAN ANSAVKNNNNE

TH2R region peptides pool

Pept 12 ANSA VKNNNNEEPSD

Pept 13 VKNN NNEEPSDKHIK

Pept 14 NNEE PSDKHIKEYLN

Pept 15 PSDK HIKEYLNKIQN

Pept 16 HIKE YLNKIQNSLST

Pept 17 YLNK IQNSLSTEWSP

Pept 18 IQNS LSTEWSPCSVT

Pept 19 LSTE WSPCSVTCGNG

TH3R/CS.T3T region peptides pool

Pept 20 WSPC SVTCGNGIQVR

Pept 21 SVTC GNGIQVRIKPG

Pept 22 GNGI QVRIKPGSANK

Pept 23 QVRI KPGSANKPKDE

Pept 24 KPGS ANKPKDELDYA

Pept 25 ANKP KDELDYANDIE

Pept 26 KDEL DYANDIEKKIC

Pept 27 DYAN DIEKKICKMEK

Pept 28 DIEK KICKMEKCSSV

Pept 29 KICK MEKCSSVFNVV

Pept 30 MEKC SSVFNVVNSSI

Pept 31 KCSS VFNVVNSSIGL

All three peptide pools were combined for the ICS assay. The pools were usedseparately for the ex vivo and plating out of the cultured ELISPOT assay.doi:10.1371/journal.pone.0025786.t001

T Cell Immunity and RTS,S/AS01E

PLoS ONE | www.plosone.org 3 October 2011 | Volume 6 | Issue 10 | e25786

Page 5: Charles Darwin University Circumsporozoite-specific T cell ... · Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01 E and Protection against P falciparumClinical

log-transformed values. Cox regression for the primary endpoint

(clinical malaria with P. falciparum density $2500/mL) was adjusted

for age at first vaccination, village, distance from the health facility,

bed net use and anti-circumsporozoite (CS) antibody levels by

dichotomizing concurrent anti-CS titers at 40 EU/mL [10].

Cellular responses were analyzed as time-varying covariates,

applying the result from the time of the most recent clinic visit.

A Bonferroni correction was subsequently calculated for the

independently significant explanatory variables. Responses were

log transformed to produce normal distributions before inclusion

in the Cox regression models. Analysis was conducted on the

According To Protocol vaccinees. STATA version 10 was used.

Results

Blood samples were processed from 407 children. Data were

acquired from 1,066 ICS assays (from three different clinic visits),

660 cultured ELISPOTs (from four different clinic visits), 780 ex

vivo ELISPOTs for IFNc (from three clinic visits) and 453 ex vivo

ELISPOTs for IL2 production (from 3 clinic visits). 56 (8%), 12

(2%) and 21 (5%) assays failed quality control criteria for positive

and negative controls for cultured, and ex vivo IFNc and ex vivo IL2

ELISPOTs, respectively. For ICS assay, the results from the

positive control were at least 100 cells per million above the

negative control for 1045 (98.0%), 1057 (99.1%) and 1055 (99.0%)

for IFNc, IL2 and TNFa, respectively.

The geometric mean responses to the negative control were 75,

165 and 159 cells per million for IFNc, IL2 and TNFa ICS results

respectively, and average responses to positive control were 3,768,

18,895 and 3,454 cells per million. The mean responses to CS

antigen vary by timepoint and by vaccination group, but the

ranges were 11 to 25, 10 to 681 and 8 to 426 for IFNc, IL2 and

TNFa, respectively. There was no variation in responses to control

by time point (p = 0.15, p = 0.15, p = 0.6) or by vaccination group

at the first timepoint post vaccination (p = 0.4, p = 0.36 and

p = 0.39). An example of the flow cytometry analysis is shown in

Figure 1.

Vaccine induced anti-CS T cell responses: ICS assaysCD4+ and CD8+ anti-CS T cell responses were detected in

both vaccination groups using ICS. There were no significant

differences between the groups pre-vaccination. Vaccination with

RTS,S/AS01E induced CD4+ but no CD8+ anti-CS T cell

responses. The strongest responses were seen for IL2 producing

CD4 T cells at one month post vaccination (a mean of 681 cells

per million, 95%CI 585–792), followed by TNFa (426 cells per

million, 95%CI 362–502), and weak IFNc responses (25 cells per

million, 95%CI 18–34) (Table 2, Figure 2). These levels

Figure 1. An example plot of FACS data acquired following intra-cellular cytokine staining is shown for negative control (mediumonly), positive control (i.e. staphylococcal enterotoxin B, SEB) and CS peptides.doi:10.1371/journal.pone.0025786.g001

T Cell Immunity and RTS,S/AS01E

PLoS ONE | www.plosone.org 4 October 2011 | Volume 6 | Issue 10 | e25786

Page 6: Charles Darwin University Circumsporozoite-specific T cell ... · Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01 E and Protection against P falciparumClinical

corresponded to 3.2 fold, 2.3 fold and 1.9 fold increases for IL2,

TNFa and IFNc, respectively.

ELISPOT assaysThree different peptide pools were used for the ELISPOT

assays, allowing a more detailed analysis of immunogenicity.

Cultured ELISPOT results were higher among RTS,S/AS01E

vaccinees than among rabies vaccinees at 1 month and 6.5 months

post vaccination, but not at 12 months. IFNc ex vivo ELISPOT

results did not vary by vaccination group at any timepoint. IL2 ex

vivo ELISPOT responses were significantly higher in RTS,S/

AS01E vaccinees at 1 month post vaccination, but not at 6.5

months (Table 2) compared with rabies vaccinees.

For both the cultured IFNc ELISPOT and ex vivo IL2

ELISPOT, the vaccine induced cellular responses were limited

to two peptide pools (i.e. TH2R and TH3R/CS.T3T pools, Table

1). No responses were detected to the third peptide pool (NANP

and conserved region peptides; Figure 3).

Table 2. Geometric means of CMI assays by clinic visit and byvaccination group.

Visit Rabies RTS,S/AS01E

Mean(95%CI) N

Mean(95%CI) N p

ICS: CD4+ve cells: IFNg

Screen 11(8–14) 197 12(9–16) 182 0.6

Vac+1 mth 13(10–18) 182 25(18–34) 170 0.01*

Vac+12 mths 11(7–15) 167 20(14–29) 168 0.009**

ICS: CD4+ve cells: IL2

Screen 103(84–127) 197 94(76–117) 182 0.52

Vac+1 mth 212(183–245) 182 681(585–792) 170 2610–13**

Vac+12 mths 10(7–14) 167 102(73–142) 168 9610–19**

ICS: CD4+ve cells: TNFa

Screen 86(69–108) 197 81(64–102) 182 0.69

Vac+1 mth 182(156–214) 182 426(362–502) 170 1610–08**

Vac+12 mths 8(6–11) 167 48(34–68) 168 6610–12**

ICS: CD8+ve cells: IFNg

Screen 12(9–17) 178 7(5–10) 175 0.028#

Vac+1 mth 19(12–28) 168 19(12–28) 156 0.99

Vac+12 mths 10(6–15) 137 11(7–17) 127 0.73

ICS: CD8+ve cells: IL2

Screen 156(128–191) 178 190(155–233) 175 0.24

Vac+1 mth 200(164–246) 168 215(175–266) 156 0.64

Vac+12 mths 9(6–13) 137 20(13–32) 127 0.004**

ICS: CD8+ve cells: TNFa

Screen 49(36–68) 178 59(43–81) 175 0.43

Vac+1 mth 133(106–167) 168 162(127–205) 156 0.26

Vac+12 mths 15(10–24) 137 16(10–25) 127 0.91

IFNg cultured ELISPOT: NANP and conserved region

Screen 27(21–35) 72 33(26–43) 70 0.22

Vac+1 mth 32(27–38) 86 28(24–32) 109 0.26

Vac+6.5 mths 29(25–35) 82 26(22–30) 122 0.34

Vac+12 mths 31(23–42) 55 27(21–36) 64 0.54

IFNg cultured ELISPOT: TH2R region

Screen 33 (26–43) 72 36 (28–48) 70 0.66

Vac+1 mth 34(25–47) 86 66(50–88) 109 3610–4**

Vac+6.5 mths 30(22–41) 83 60(47–78) 122 2610–4**

Vac+12 mths 26(20–34) 55 39(30–51) 64 0.023*

IFNg cultured ELISPOT: TH3R/ CS.T3T region

Screen 31 (23–41) 72 37 (28–48) 70 0.38

Vac+1 mth 30(23–40) 86 55(43–70) 109 3610–4**

Vac+6.5 mths 32(23–44) 83 58(45–76) 122 0.003**

Vac+12 mths 33(24–46) 55 36(26–48) 64 0.79

IFNg cultured ELISPOT: All CS peptides summed

Screen 75 (58–97) 72 90 (69–117) 70 0.7

Vac+1 mth 88(66–117) 86 151(117–195) 109 0.002**

Vac+6.5 mths 81(59–110) 82 145(113–187) 122 0.002**

Vac+12 mths 83(59–115) 55 104(76–141) 64 0.33

IFNg ex vivo ELISPOT: NANP and conserved region

Screen 15(13–17) 152 14(12–16) 137 0.59

Vac+1 mth 15(13–18) 100 16(13–19) 104 0.83

Visit Rabies RTS,S/AS01E

Mean(95%CI) N

Mean(95%CI) N p

Vac+6.5 mths 13(12–14) 145 12(11–13) 142 0.21

IFNg ex vivo ELISPOT: TH2R region

Screen 15(13–17) 152 14(12–16) 137 0.56

Vac+1 mth 16(13–19) 100 18(15–22) 104 0.23

Vac+6.5 mths 14(13–16) 145 14(13–17) 142 0.96

IFNg ex vivo ELISPOT: TH3R/ CS.T3T region

Screen 18(15–20) 152 17(15–20) 137 0.79

Vac+1 mth 19(15–23) 100 21(17–25) 104 0.43

Vac+6.5 mths 13(11–15) 144 15(13–17) 141 0.23

IFNg ex vivo ELISPOT: All CS peptides summed

Screen 40(35–46) 152 39(34–45) 137 0.69

Vac+1 mth 44(36–54) 100 48(40–59) 104 0.5

Vac+6.5 mths 35(31–40) 144 35(31–40) 141 0.94

IL2 ex vivo ELISPOT: NANP and conserved region

Screen 15(13–18) 107 15(12–18) 89 0.8

Vac+1 mth 12(10–14) 62 14(12–17) 56 0.11

Vac+6.5 mths 15(12–20) 76 15(11–21) 63 0.98

IL2 ex vivo ELISPOT: TH2R region

Screen 16(13–19) 107 16(13–20) 89 0.93

Vac+1 mth 13(10–17) 62 21(16–28) 56 0.003**

Vac+6.5 mths 18(14–23) 76 17(13–22) 63 0.76

IL2 ex vivo ELISPOT: TH3R/ CS.T3T region

Screen 17(15–21) 107 17(14–21) 89 0.9

Vac+1 mth 15(12–19) 62 24(19–31) 56 0.003**

Vac+6.5 mths 18(13–24) 76 29(20–41) 62 0.022*

IL2 ex vivo ELISPOT: All peptides summed

Screen 44(37–53) 107 42(34–51) 89 0.71

Vac+1 mth 35(27–45) 62 56(44–73) 56 0.002**

Vac+6.5 mths 45(34–60) 76 53(39–72) 62 0.43

*for p,0.05 or ** for p,0.005 where Mean for Rabies group , Mean for RTS,S/AS01E group. # for p,0.05 or ## for p,0.005 where Mean for Rabies group .

Mean for RTS,S/AS01E group.doi:10.1371/journal.pone.0025786.t002

Table 2. Cont.

T Cell Immunity and RTS,S/AS01E

PLoS ONE | www.plosone.org 5 October 2011 | Volume 6 | Issue 10 | e25786

Page 7: Charles Darwin University Circumsporozoite-specific T cell ... · Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01 E and Protection against P falciparumClinical

Time course of responses (ICS assays)The frequencies of IL2, TNFa and IFNc producing CD4+ T

cells by ICS was significantly higher at one month after the final

vaccination with RTS,S/AS01E compared with pre-vaccination

levels (p,0.0001, p,0.0001, p = 0.0006, respectively). There was

then a fall in responses between 1 month and 12 months post

vaccination, falling to pre-vaccination levels for IL2 (p,0.0001)

and TNFa (p,0.0001). IFNc producing CD4+ T cells remained

above pre-vaccination levels, albeit at low frequency throughout.

However, there was an even more pronounced fall in CD4+ T cell

responses among control vaccinees (Table 2), and so RTS,S/

AS01E vaccinees had substantially higher T cell responses than

control vaccinees at 12 months post vaccination (p,0.0001 for

TNFa and IL2, p = 0.009 for IFNc).

Inter assay correlationsThere were strong correlations between the different cytokines

detected by ICS, and also between IL2/IFNc ELISPOT results

one month after vaccination with RTS,S/AS01E (Table 3).

Cultured ELISPOTs were significantly associated with ICS results,

but not with ex vivo ELISPOT results (IFNc or IL2). Antibody titres

were associated with all the cellular assays except ex vivo IFNcELISPOTs (Table 3).

Correlates of immunityAfter vaccination with RTS,S/AS01E, an increasing frequency

of TNFa producing, CS-specific CD4+ cells detected using ICS

was associated with a reduced risk of clinical malaria (HR = 0.64

for each 10 fold increase in the frequency of CD4+ TNFa+ T cells,

95%CI 0.49–0.86, p = 0.002). On ICS, IFNc production by CS-

specific CD4+ T cells was associated with a reduced risk of clinical

malaria of borderline significance (p = 0.07, Table 4). TNFa and

IFNc producing CS-specific CD4+ T cells were at much lower

frequencies among control vaccinees, but nevertheless were

associated with reduced risks of clinical malaria of borderline

significance. When data from both RTS,S/AS01E vaccinees and

control vaccinees were combined (with adjusting for vaccination

group), the overall hazard ratios were 0.74 (95%CI 0.62–0.89,

p = 0.001) and 0.79 (95%CI 0.67–0.94, p = 0.007) for TNFa and

IFNc, respectively. On Bonferroni adjustment, these p values were

0.018 and 0.13, respectively. Similar results were observed when

adjusting for anti-CSP antibody titres as a continuous variable

(HR = 0.75, 95%CI 0.62–0.91, p = 0.003 with p = 0.054 after

Bonferroni adjustment, and HR = 0.81, 95%CI 0.68–0.95,

p = 0.01 with p = 0.13 after Bonferroni adjustment for TNFaCD4+ T cells and IFNc CD4+ T cells, respectively).

In order to display the effect graphically, the cellular responses

were split by tertile (Figure 4). The middle tertiles for TNFa are at

intermediate risk, suggesting a continuous change in risk as the

frequency of TNFa cells increases rather than a threshold effect.

When the frequencies of TNFa producing CD4+ T cells and

IFNc producing CD4+ T cells were combined in the same model,

TNFa CD4+ T cell frequency was the independent factor (i.e.

HR = 0.76, 95%CI 0.64–0.89, p = 0.001 compared with

HR = 0.84, 95%CI 0.71–1.00, p = 0.050 for the frequency of

IFNc producing CD4+ T cells). An interaction term generated by

multiplying the frequency of TNFa CD4+ T cells by antibody

concentrations was not significant in determining risk (HR = 0.79,

95%CI 0.51–1.2, p = 0.29).

On applying the fourth of the Prentice criteria, we found that

vaccination group was still an independent predictor of clinical malaria

risk in a multivariable model including CD4+ TNFa+ cells

(HR = 0.69, 95%CI 0.48–0.97, p = 0.036). In other words, only 42%

of the effect of vaccination could be accounted for by CD4+ TNFa+cells. However, when anti-CS titers were added to the model the effect

of vaccine became non-significant (HR = 0.93, 95%CI 0.62–1.42,

p = 0.76, i.e. 87% of the effect of vaccination was accounted for).

Hence, while neither CD4+ TNFa+ cells nor anti-CS antibodies

alone accounted for all of the effect of vaccination with RTS,S/

AS01E on clinical malaria risk, the combination of CD4+ TNFa+T cells and anti-CS antibodies together could account for all of the

statistical effect of vaccination.

Discussion

Vaccination with RTS,S/AS01E induced circumsporozoite

protein (CS) specific T cell responses in 5–17 month-old children

living in a malaria endemic area. The frequency of CD4+ TNFa+ T

cells on ICS was associated with protection from clinical malaria.

Although the use of 15-mer peptides may have been sub-optimal to

demonstrate CD8+ T cell responses, we did in fact identify both

CD4+ and CD8+ responses above negative control conditions for

both RTS,S/AS01E and control vaccinees. However, the CD8+responses were apparently not induced by vaccination, and

presumably are the result of exposure to malaria parasites [32].

If TNFa producing CD4+ T cells are causally related to

protection, they should be associated with protection whether they

are acquired by vaccination or by natural exposure to malaria

Table 3. Inter-assay Correlation coefficients of CMI assays at 1 month post vaccination with RTS,S/AS01E.

CD4+ IFNc CD4+ IL2 CD4+ TNFa Antibody (CS) Cultured IFNc IFNc ELISPOT IL2 ELISPOT

CD4+ IFNc 1

CD4+ IL2 0.38*** 1

CD4+ TNFa 0.32*** 0.66*** 1

Antibody (CS) 0.14*** 0.35*** 0.26*** 1

Cultured IFNc 0.14** 0.15** 0.18*** 0.22*** 1

IFNc ELISPOT 20.05 20.05 0.02 0.03 20.02 1

IL2 ELISPOT 0 0.01 0.15* 0.11* 20.06 0.31*** 1

* = p,0.05.** = p,0.001.*** = p,0.0001.doi:10.1371/journal.pone.0025786.t003

T Cell Immunity and RTS,S/AS01E

PLoS ONE | www.plosone.org 6 October 2011 | Volume 6 | Issue 10 | e25786

Page 8: Charles Darwin University Circumsporozoite-specific T cell ... · Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01 E and Protection against P falciparumClinical

Figure 2. The time course of anti-CS CD4+ ICS responses and summed ELISPOT responses is shown per time point for RTS,S/AS01E

and control vaccination groups. * indicates p,0.05 and ** indicates p,0.005.doi:10.1371/journal.pone.0025786.g002

Figure 3. ELISPOT responses are shown for the individual stimulating peptide pools at 1 month post vaccination with RTS,S/AS01E.doi:10.1371/journal.pone.0025786.g003

T Cell Immunity and RTS,S/AS01E

PLoS ONE | www.plosone.org 7 October 2011 | Volume 6 | Issue 10 | e25786

Page 9: Charles Darwin University Circumsporozoite-specific T cell ... · Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01 E and Protection against P falciparumClinical

parasites. Indeed, a borderline statistical correlation between

TNFa+ CD4+ T cells and a reduced risk of clinical malaria was

observed among control vaccinees. However, the overall level of

protection afforded by T cells among RTS,S/AS01E vaccinees was

greater, since the vaccinated children had 2–3 fold more TNFa+CD4+ T cells at 1 month and 8–10 fold more at 12 months post

vaccination. The frequency of TNFa_CD4+ T cells at 12 months

post vaccination was below screening (i.e. pre-vaccination) levels,

despite the fact that clinical protection was sustained at 15 months

[10]. Since clinical protection is determined by contemporaneous

comparison with control vaccinees, it is therefore theoretically

possible that the relative difference between RTS,S/AS01E

vaccinees and control vaccines is relevant despite this fall.

T cell responses to CSP rose between screening and 1 month

post-vaccination, and then fell to lower levels at 12 months post

vaccination. This was seen in both ELISPOT and ICS studies, and

could not be explained by greater background responses (which did

not change over time and were subtracted from antigen-specific

responses), or by non-specific responses detected in changing

positive controls over time. The temporary increase in CSP specific

responses parallels the increase and decrease in antibodies to blood

stage antigens seen in the same children [33]. Antibody responses

made by young children to blood stage antigens are often short-lived

[34], and may reflect short-term changes in exposure [35]. Taking

together the antibody data, the consistent pattern in ICS and

ELISPOT studies, the stability of positive and negative control

responses, and the accounting for background reactivity by

subtracting negative control responses from antigen-specific re-

sponses, show no significant variation over time for positive and

negative controls, we conclude that the T cell responses are raised

by exposure to malaria during the transmission season, but are

short-lived and therefore not sustained once exposure falls.

We did not identify an association between protection and T

cell responses detected by the cultured IFNc assay, as previously

reported [20]. However, we tested pools of peptides rather than

individual peptides, and the previously reported association was

specific to the CS.T3T peptide. The CS.T3T peptide was

contained in a pool of TH3R/CS.T3T peptides. In other studies,

the CS.T3T peptide accounted for more than half of the overall

response seen in the TH3R/CS.T3T peptide pool [36].

Furthermore, we identified very little IFNc production in our

study. Previous studies showing marked IFNc production have

been done in adults [18], and IFNc production may be suppressed

in children in malaria endemic areas [37].

Ex vivo ELISPOT studies did not correlate with ICS studies for

the same cytokines, even though both use overnight stimulations,

and ICS results were 10-fold higher than ELISPOT results. This

may be partially explained by measuring ELISPOT assays per

million PBMC, whereas ICS is measured per million CD4+ T

cells. However, ex vivo and cultured ELISPOTs identify different

Table 4. The hazard ratio from Cox regression models (with 95% CI) for the outcome clinical malaria by CMI assays.

Both datasets Rabies Vaccinees RTS,S/AS01E Vaccinees

Assay HR (95%CI) p HR (95%CI) p HR (95%CI) p

ICS: CD4 cells

IFNc 0.79(0.67–0.94) 0.007 0.81(0.66–1.01) 0.058 0.77(0.59–1.02) 0.07

IL2 0.9(0.76–1.07) 0.23 0.97(0.78–1.22) 0.81 0.77(0.55–1.08) 0.13

TNFa 0.74(0.62–0.89) 0.001 0.8(0.62–1.03) 0.084 0.64(0.49–0.86) 0.002

ICS: CD8 cells

IFNc 1.07(0.91–1.25) 0.43 1.13(0.93–1.37) 0.21 0.93(0.68–1.27) 0.65

IL2 0.85(0.69–1.05) 0.13 0.92(0.7–1.21) 0.56 0.79(0.56–1.12) 0.19

TNFa 0.87(0.72–1.04) 0.12 0.83(0.66–1.05) 0.11 0.93(0.68–1.28) 0.66

IFNc cultured ELISPOT

NANP 1.01(0.65–1.57) 0.95 0.9(0.54–1.52) 0.7 1.08(0.44–2.68) 0.87

TH2R 0.76(0.51–1.14) 0.18 0.67(0.3–1.52) 0.34 0.79(0.49–1.27) 0.33

TH3R 0.94(0.67–1.32) 0.72 0.99(0.61–1.61) 0.97 0.86(0.53–1.39) 0.54

Sum 0.95(0.67–1.34) 0.77 0.92(0.54–1.57) 0.77 0.92(0.58–1.47) 0.73

IFNc ex vivo ELISPOT

NANP 1.61(1.01–2.55) 0.044 1.54(0.87–2.72) 0.14 1.38(0.62–3.08) 0.44

TH2R 1(0.59–1.69) 1 0.9(0.44–1.85) 0.78 1.1(0.49–2.45) 0.81

TH3R 1.62(1.04–2.52) 0.032 1.57(0.82–2.99) 0.17 1.4(0.75–2.64) 0.29

Sum 1.35(0.86–2.12) 0.2 1.32(0.73–2.4) 0.35 1.23(0.61–2.47) 0.57

IL2 ex vivo ELISPOT

NANP 1.18(0.56–2.51) 0.67 1(0.29–3.39) 0.99 1.38(0.46–4.17) 0.57

TH2R 0.57(0.23–1.45) 0.24 0.49(0.13–1.79) 0.28 0.81(0.2–3.2) 0.76

TH3R 0.94(0.45–1.97) 0.87 0.73(0.21–2.49) 0.61 0.97(0.39–2.42) 0.94

Sum 0.83(0.38–1.83) 0.65 0.81(0.25–2.56) 0.72 0.78(0.23–2.62) 0.69

HR = Hazard Ratio for each log (i.e. ten-fold) increase in frequency of T cells. Confidence intervals are 5–95%. HRs are adjusted by anti-CS antibody titre (in 2 groups),age, area of residence, ITN use and distance from the dispensary. NANP = NANP and conserved region peptides pool, TH2R = TH2R region peptides pool, TH3R = TH3Rand CS.T3T region peptide pool, Sum = all three peptide pools summed.doi:10.1371/journal.pone.0025786.t004

T Cell Immunity and RTS,S/AS01E

PLoS ONE | www.plosone.org 8 October 2011 | Volume 6 | Issue 10 | e25786

Page 10: Charles Darwin University Circumsporozoite-specific T cell ... · Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01 E and Protection against P falciparumClinical

cell populations, the latter more closely reflecting a central

memory phenotype [38,39,40]. Hence it is possible that ICS and

cultured ELISPOT identify central memory cells, but ex vivo

ELISPOTs identify effector phenotypes. ELISPOT assays failing

internal control standards were excluded, but similar control

standards were not pre-defined for ICS data. However, this

applied to a minority of assays (2% and 5%, respectively for ex vivo

IFNc and IL2 ELISPOTS) which did not correlate with ICS data,

and 8% for cultured ELISPOTS, which did correlate. Hence it

seems unlikely that lack of quality control standards for ICS

explained the lack of correlation.

The CD4+ T cell response associated with protection in our

analysis (i.e. TNFa production) were at a low frequency (mean 426

cells per million CD4 cells at peak). Higher frequency responses

have been required to achieve protection in sporozoite challenge

studies [7]. However, the antibody concentrations associated with

protection are also higher in sporozoite challenge studies [7,10].

These differences in outcome may be explained by the greater

sporozoite inoculum used in challenge studies compared with

exposure in the field [41]. The IFNc response that was apparently

associated with protection in our study was very low frequency and

barely above the limit of detection (25 cells per million), and the

apparent association is likely to reflect the association between

IFNc and TNFa rather than an independent effect. We were

unable to assess polyfunctionality in the present study, because the

high number of samples (n = 1066, with three conditions per

sample) required an automated gating strategy using FACSDiva

software (BD Biosciences). The FACSdiva analysis used for this

study did not include Boolean gates in order to obtain

polyfunctional T cell data. Further analysis to determine

polyfunctionality in this large dataset is ongoing.

Multiple comparisons (i.e. 18) have been undertaken to identify

the association between TNFa producing CD4+ T cells and

protection from clinical malaria. However, the association was

highly significant (p = 0.001) and remains significant after a

Bonferroni correction (p = 0.018).

The Prentice criteria have been proposed as a way of qualifying

surrogate endpoints [42,43] and include four criteria [30]. We

found that the combination of anti-CS titers and TNFa producing

CD4+ T cells met all the criteria (i.e. vaccination was associated

with protection; anti-CS titers and CD4+ TNFa+ T cells were

both independently associated with vaccination; were both

Figure 4. Survival plots with time to first episode of clinical malaria plotted for RTS,S/AS01E (left columns) and control vaccinees(left and right columns) according to tertile of CD4+, TNFa responses (top row), CD4+ IFNc responses (middle row) and IFNc ex vivoELISPOT responses to TH3R/CS.T3T peptides pool (lower row). Where more than one third of responses were at the lower limit of detection,the lower two tertiles are combined (and hence only 2 tertiles are displayed on some plots). For CD4+ TNFa+ responses, the tertiles were 1 to 154(lower), 155 to 407 (middle) and 408 to 28,840 (upper) cells per million for RTS,S/AS01E vaccinees, and 1 to 26 (lower), 27 to 165 (middle) and 166 to10,000 (upper) cells per million for control vaccinees. For CD+ IFNc+ responses the tertiles were 1 to 12 (lower), 13 to 66 (middle) and 67 to 8,320(upper) cells per million for RTS,S/AS01E vaccinees, and 1 to 40 (lower) and 41 to 5,980 (upper) cells per million for rabies vaccinees. The time point ‘‘0months’’ refers to the time of a blood draw. Cellular responses were analyzed as time-varying covariates, where the effect of cellular responses fromall available blood draws was related to clinical malaria episodes during the period of monitoring after each measurement. Therefore, each RTS,Svaccinee could contribute to 2 periods of monitoring. These three assays were selected for the figure because significant associations on Coxregression were seen (Table 4).doi:10.1371/journal.pone.0025786.g004

T Cell Immunity and RTS,S/AS01E

PLoS ONE | www.plosone.org 9 October 2011 | Volume 6 | Issue 10 | e25786

Page 11: Charles Darwin University Circumsporozoite-specific T cell ... · Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01 E and Protection against P falciparumClinical

independently associated with protection; and the combination of

anti-CS titers and CD4+ TNFa+ T cells, but not either alone,

could account for the effect of vaccination in multi-variable Cox

regression analysis). We found no significant interaction between

anti-CS titers and TNFa+ T cells.

Microheterogeneity of malaria exposure has been observed in

Kilifi [44], and may confound the association between antibodies

to blood stage malaria antigens and the risk of malaria [33].

However, this is unlikely to explain the association between CD4+TNFa+ T cells and protection from clinical malaria for two

reasons: The direction of confounding was in the opposite

direction in this cohort (i.e. microheterogeneity led to a

confounded association between increasing antibody levels and

increasing risk of malaria rather than protection), and the

association with protection is more marked in RTS,S/AS01E

vaccinees rather than control vaccinees.

There is strong evidence that anti-CS antibodies inhibit

sporozoite invasion [45], supporting a causal relationship, and

TNFa may reduce the parasite’s intrahepatocytic development

[13,14]. However, it is possible that the frequency of CD4+TNFa+ T cells is associated with another causal mediator of

immunity (for instance better quality antibody responses, en-

hanced T cell memory or polyfunctionality). These further

characterizations of the immune response should now be a

priority, since establishing an immunological surrogate endpoint

will accelerate the development of candidate malaria vaccines,

inform monitoring the persistence of immune responses and

inform the timing of a booster dose.

Supporting Information

Supporting Information S1

(PPT)

Acknowledgments

We are grateful to the parents of participants and village and district

authorities for their cooperation; the Data and safety monitoring board,

chaired by Prof. Malcolm Molyneux, and the local safety monitor, Dr. Jay

Berkley in Kilifi; Edna Ogada, Juliana Wambua (site coordinator), and

Dorothy Mwachiro (community liaison officer) for providing support in

Kilifi; and the staff of the Malaria Project Team at GlaxoSmithKline — in

particular, Mickael Mestre (technical assistance), Olivier Jauniaux

(technical assistance), Robbert Van-der-Most and Lode Schuerman for

pre-submission reviews of the manuscript, Nathalie Annez, Sarah Benns

(professional writer), Srilakshmi Pranesh, Marie-Sylvie Remacle (profes-

sional writer) and Laurence Vigneron.

Author Contributions

Conceived and designed the experiments: PM JV ML AL TV BS KM JC

PB. Performed the experiments: AO PM JM DK MJ OK EJ PB. Analyzed

the data: AO ML PB. Wrote the paper: AO JV AL JC PB.

References

1. Ballou WR (2009) The development of the RTS,S malaria vaccine candidate:

challenges and lessons. Parasite Immunol 31: 492–500.

2. Lell B, Agnandji S, von Glasenapp I, Haertle S, Oyakhiromen S, et al. (2009) A

randomized trial assessing the safety and immunogenicity of AS01 and AS02 adjuvanted

RTS,S malaria vaccine candidates in children in Gabon. PLoS One 4: e7611.

3. Bojang KA, Olodude F, Pinder M, Ofori-Anyinam O, Vigneron L, et al. (2005)

Safety and immunogenicty of RTS,S/AS02A candidate malaria vaccine in

Gambian children. Vaccine 23: 4148–4157.

4. Bojang KA, Milligan PJ, Pinder M, Vigneron L, Alloueche A, et al. (2001)

Efficacy of RTS,S/AS02 malaria vaccine against Plasmodium falciparum

infection in semi-immune adult men in The Gambia: a randomised trial. Lancet

358: 1927–1934.

5. Doherty JF, Pinder M, Tornieporth N, Carton C, Vigneron L, et al. (1999) A

phase I safety and immunogenicity trial with the candidate malaria vaccine

RTS,S/SBAS2 in semi-immune adults in The Gambia. Am J Trop Med Hyg

61: 865–868.

6. Stoute JA, Slaoui M, Heppner DG, Momin P, Kester KE, et al. (1997) A

preliminary evaluation of a recombinant circumsporozoite protein vaccine

against Plasmodium falciparum malaria. RTS,S Malaria Vaccine Evaluation

Group. N Engl J Med 336: 86–91.

7. Kester KE, Cummings JF, Ofori-Anyinam O, Ockenhouse CF, Krzych U, et al.

(2009) Randomized, double-blind, phase 2a trial of falciparum malaria vaccines

RTS,S/AS01B and RTS,S/AS02A in malaria-naive adults: safety, efficacy, and

immunologic associates of protection. J Infect Dis 200: 337–346.

8. Guinovart C, Aponte JJ, Sacarlal J, Aide P, Leach A, et al. (2009) Insights into

long-lasting protection induced by RTS,S/AS02A malaria vaccine: further

results from a phase IIb trial in Mozambican children. PLoS One 4: e5165.

9. Alonso PL, Sacarlal J, Aponte JJ, Leach A, Macete E, et al. (2004) Efficacy of the

RTS,S/AS02A vaccine against Plasmodium falciparum infection and disease in

young African children: randomised controlled trial. Lancet 364: 1411–1420.

10. Olotu A, Lusingu J, Leach A, Lievens M, Vekemans J, et al. (2011) Efficacy of

RTS,S/AS01E malaria vaccine and exploratory analysis on anti-circumspor-

ozoite antibody titres and protection in children aged 5-17 months in Kenya and

Tanzania: a randomised controlled trial. Lancet Infect Dis 11: 102–109.

11. Wang R, Charoenvit Y, Corradin G, De La Vega P, Franke ED, et al. (1996)

Protection against malaria by Plasmodium yoelii sporozoite surface protein 2

linear peptide induction of CD4+ T cell- and IFN-gamma-dependent

elimination of infected hepatocytes. J Immunol 157: 4061–4067.

12. McConkey SJ, Reece WH, Moorthy VS, Webster D, Dunachie S, et al. (2003)

Enhanced T-cell immunogenicity of plasmid DNA vaccines boosted by

recombinant modified vaccinia virus Ankara in humans. Nat Med 9: 729–735.

13. Butler NS, Schmidt NW, Harty JT (2010) Differential effector pathways regulate

memory CD8 T cell immunity against Plasmodium berghei versus P. yoelii

sporozoites. J Immunol 184: 2528–2538.

14. Depinay N, Franetich JF, Gruner AC, Mauduit M, Chavatte JM, et al. (2011)

Inhibitory effect of TNF-alpha on malaria pre-erythrocytic stage development:

influence of host hepatocyte/parasite combinations. PLoS ONE 6: e17464.

15. Doolan DL, Hoffman SL (2000) The complexity of protective immunity against

liver-stage malaria. J Immunol 165: 1453–1462.

16. Casares S, Brumeanu TD, Richie TL (2010) The RTS,S malaria vaccine.

Vaccine 28: 4880–4894.

17. Moorthy VS, Ballou WR (2009) Immunological mechanisms underlying

protection mediated by RTS,S: a review of the available data. Malar J 8: 312.

18. Lalvani A, Moris P, Voss G, Pathan AA, Kester KE, et al. (1999) Potent

induction of focused Th1-type cellular and humoral immune responses by

RTS,S/SBAS2, a recombinant Plasmodium falciparum malaria vaccine. J Infect

Dis 180: 1656–1664.

19. Stoute JA, Kester KE, Krzych U, Wellde BT, Hall T, et al. (1998) Long-term

efficacy and immune responses following immunization with the RTS,S malaria

vaccine. J Infect Dis 178: 1139–1144.

20. Reece WH, Pinder M, Gothard PK, Milligan P, Bojang K, et al. (2004) A

CD4(+) T-cell immune response to a conserved epitope in the circumsporozoite

protein correlates with protection from natural Plasmodium falciparum infection

and disease. Nat Med 10: 406–410.

21. Pinder M, Reece WH, Plebanski M, Akinwunmi P, Flanagan KL, et al. (2004)

Cellular immunity induced by the recombinant Plasmodium falciparum malaria

vaccine, RTS,S/AS02, in semi-immune adults in The Gambia. Clin Exp

Immunol 135: 286–293.

22. Sun P, Schwenk R, White K, Stoute JA, Cohen J, et al. (2003) Protective

immunity induced with malaria vaccine, RTS,S, is linked to Plasmodium

falciparum circumsporozoite protein-specific CD4+ and CD8+ T cells

producing IFN-gamma. J Immunol 171: 6961–6967.

23. Barbosa A, Naniche D, Aponte JJ, Manaca MN, Mandomando I, et al. (2009)

Plasmodium falciparum-specific cellular immune responses after immunization

with the RTS,S/AS02D candidate malaria vaccine in infants living in an area of

high endemicity in Mozambique. Infect Immun 77: 4502–4509.

24. Olotu AI, Fegan G, Bejon P (2010) Further analysis of correlates of protection

from a phase 2a trial of the falciparum malaria vaccines RTS,S/AS01B and

RTS,S/AS02A in malaria-naive adults. J Infect Dis 201: 970–971.

25. Bejon P, Lusingu J, Olotu A, Leach A, Lievens M, et al. (2008) Efficacy of

RTS,S/AS01E vaccine against malaria in children 5 to 17 months of age. N

Engl J Med 359: 2521–2532.

26. Ansong D, Asante KP, Vekemans J, Owusu SK, Owusu R, et al. (2011) T cell

responses to the RTS,S/AS01(E) and RTS,S/AS02(D) malaria candidate

vaccines administered according to different schedules to Ghanaian children.

PLoS ONE 6: e18891.

27. Agnandji ST, Fendel R, Mestre M, Janssens M, Vekemans J, et al. (2011)

Induction of Plasmodium falciparum-specific CD4+ T cells and memory B cells

T Cell Immunity and RTS,S/AS01E

PLoS ONE | www.plosone.org 10 October 2011 | Volume 6 | Issue 10 | e25786

Page 12: Charles Darwin University Circumsporozoite-specific T cell ... · Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01 E and Protection against P falciparumClinical

in Gabonese children vaccinated with RTS,S/AS01(E) and RTS,S/AS02(D).

PLoS ONE 6: e18559.

28. Plotkin SA (2008) Vaccines: correlates of vaccine-induced immunity. Clin Infect

Dis 47: 401–409.

29. Qin L, Gilbert PB, Corey L, McElrath MJ, Self SG (2007) A framework for

assessing immunological correlates of protection in vaccine trials. J Infect Dis

196: 1304–1312.

30. Prentice RL (1989) Surrogate endpoints in clinical trials: definition and

operational criteria. Stat Med 8: 431–440.

31. Macete EV, Sacarlal J, Aponte JJ, Leach A, Navia MM, et al. (2007) Evaluation

of two formulations of adjuvanted RTS, S malaria vaccine in children aged 3 to

5 years living in a malaria-endemic region of Mozambique: a Phase I/IIb

randomized double-blind bridging trial. Trials 8: 11.

32. Flanagan KL, Lee EA, Gravenor MB, Reece WH, Urban BC, et al. (2001)

Unique T cell effector functions elicited by Plasmodium falciparum epitopes in

malaria-exposed Africans tested by three T cell assays. J Immunol 167:

4729–4737.

33. Bejon P, Cook J, Bergmann-Leitner E, Olotu A, Lusingu J, et al. (2011) Effect of

the Pre-erythrocytic Candidate Malaria Vaccine RTS,S/AS01E on Blood Stage

Immunity in Young Children. J Infect Dis 204: 9–18.

34. Kinyanjui SM, Bull P, Newbold CI, Marsh K (2003) Kinetics of antibody

responses to Plasmodium falciparum-infected erythrocyte variant surface

antigens. J Infect Dis 187: 667–674.

35. Bejon P, Turner L, Lavstsen T, Cham G, Olotu A, et al. (2011) Serological

Evidence of Discrete Spatial Clusters of Plasmodium falciparum Parasites. PLoS

ONE 6: e21711.

36. Dunachie SJ, Walther M, Vuola JM, Webster DP, Keating SM, et al. (2006) A

clinical trial of prime-boost immunisation with the candidate malaria vaccines

RTS,S/AS02A and MVA-CS. Vaccine 24: 2850–2859.

37. Bejon P, Mwacharo J, Kai O, Todryk S, Keating S, et al. (2007) The induction

and persistence of T cell IFN-gamma responses after vaccination or naturalexposure is suppressed by Plasmodium falciparum. J Immunol 179: 4193–4201.

38. Godkin AJ, Thomas HC, Openshaw PJ (2002) Evolution of epitope-specific

memory CD4(+) T cells after clearance of hepatitis C virus. J Immunol 169:2210–2214.

39. Keating SM, Bejon P, Berthoud T, Vuola JM, Todryk S, et al. (2005) DurableHuman Memory T Cells Quantifiable by Cultured Enzyme-Linked Immuno-

spot Assays Are Induced by Heterologous Prime Boost Immunization and

Correlate with Protection against Malaria. J Immunol 175: 5675–5680.40. Todryk SM, Pathan AA, Keating S, Porter DW, Berthoud T, et al. (2009) The

relationship between human effector and memory T cells measured by ex vivo andcultured ELISPOT following recent and distal priming. Immunology 128: 83–91.

41. Bejon P, Andrews L, Andersen RF, Dunachie S, Webster D, et al. (2005)Calculation of liver-to-blood inocula, parasite growth rates, and preerythrocytic

vaccine efficacy, from serial quantitative polymerase chain reaction studies of

volunteers challenged with malaria sporozoites. J Infect Dis 191: 619–626.42. Kohberger RC, Jemiolo D, Noriega F (2008) Prediction of pertussis vaccine

efficacy using a correlates of protection model. Vaccine 26: 3516–3521.43. Ray ME, Bae K, Hussain MH, Hanks GE, Shipley WU, et al. (2009) Potential

surrogate endpoints for prostate cancer survival: analysis of a phase III

randomized trial. J Natl Cancer Inst 101: 228–236.44. Bejon P, Williams TN, Liljander A, Noor AM, Wambua J, et al. (2010) Stable

and unstable malaria hotspots in longitudinal cohort studies in Kenya. Plos Med7: e1000304.

45. Hollingdale MR, Appiah A, Leland P, do Rosario VE, Mazier D, et al. (1990)Activity of human volunteer sera to candidate Plasmodium falciparum

circumsporozoite protein vaccines in the inhibition of sporozoite invasion assay

of human hepatoma cells and hepatocytes. Trans R Soc Trop Med Hyg 84:325–329.

T Cell Immunity and RTS,S/AS01E

PLoS ONE | www.plosone.org 11 October 2011 | Volume 6 | Issue 10 | e25786